14 research outputs found

    Regulation of eosinophilia and allergic airway inflammation by the glycan-binding protein galectin-1

    Get PDF
    Galectin-1 (Gal-1), a glycan-binding protein with broad antiinflammatory activities, functions as a proresolving mediator in autoimmune and chronic inflammatory disorders. However, its role in allergic airway inflammation has not yet been elucidated. We evaluated the effects of Gal-1 on eosinophil function and its role in a mouse model of allergic asthma. Allergen exposure resulted in airway recruitment of Gal-1-expressing inflammatory cells, including eosinophils, as well as increased Gal-1 in extracellular spaces in the lungs. In vitro, extracellular Gal-1 exerted divergent effects on eosinophils that were N-glycan- And dose-dependent. At concentrations ≤0.25 μM, Gal-1 increased eosinophil adhesion to vascular cell adhesion molecule-1, caused redistribution of integrin CD49d to the periphery and cell clustering, but inhibited ERK(1/2) activation and eotaxin-1-induced migration. Exposure to concentrations ≥1 μM resulted in ERK(1/2)- dependent apoptosis and disruption of the F- Actin cytoskeleton. At lower concentrations, Gal-1 did not alter expression of adhesion molecules (CD49d, CD18, CD11a, CD11b, L-selectin) or of the chemokine receptor CCR3, but decreased CD49d and CCR3 was observed in eosinophils treated with higher concentrations of this lectin. In vivo, allergen-challenged Gal-1-deficient mice exhibited increased recruitment of eosinophils and CD3+ T lymphocytes in the airways as well as elevated peripheral blood and bone marrow eosinophils relative to corresponding WT mice. Further, these mice had an increased propensity to develop airway hyperresponsiveness and displayed significantly elevated levels of TNF-α in lung tissue. This study suggests that Gal-1 can limit eosinophil recruitment to allergic airways and suppresses airway inflammation by inhibiting cell migration and promoting eosinophil apoptosis.Fil: Ge, Xiao Na. University of Minnesota; Estados UnidosFil: Ha, Sung Gil. University of Minnesota; Estados UnidosFil: Greenberg, Yana G.. University of Minnesota; Estados UnidosFil: Rao, Amrita. University of Minnesota; Estados UnidosFil: Bastan, Idil. University of Minnesota; Estados UnidosFil: Blidner, Ada Gabriela. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; ArgentinaFil: Rao, Savita P.. University of Minnesota; Estados UnidosFil: Rabinovich, Gabriel Adrián. Universidad de Buenos Aires; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; ArgentinaFil: Sriramarao, P.. University of Minnesota; Estados Unido

    Immunotherapy in cancer. Current prospects, challenges and new horizons

    Get PDF
    Recent understanding of the mechanisms that control immune system homeostasis and orchestrate antitumor responses has prompted the development of novel immunotherapeutic modalities. These include antibodies that target immune checkpoints such as PD-1/PD-L1 and CTLA-4, agonistic antibodies of costimulatory molecules such as CD137 and OX-40 and the adoptive transfer of genetically-modified antitumor T cells. However, a large number of patients do not respond to these therapies and develop resistance as a result of activation of compensatory circuits. Rational combination of immunotherapeutic modalities will help overcome resistance and will increase the number of patients who will benefit from these treatments. Moreover, identification of predictive biomarkers will allow selection of patients responding to these treatments. Emerging clinical trials and pre-clinical studies have shown exciting results anticipating new horizons in the design and implementation of cancer immunotherapeutic modalities

    Galectin-1 fosters an immunosuppressive microenvironment in colorectal cancer by reprogramming CD8⁺ regulatory T cells

    Get PDF
    Colorectal cancer (CRC) represents the third most common malignancy and the second leading cause of cancer-related deaths worldwide. Although immunotherapy has taken center stage in mainstream oncology, it has shown limited clinical efficacy in CRC, generating an urgent need for discovery of new biomarkers and potential therapeutic targets. Galectin-1 (Gal-1), an endogenous glycan-binding protein, induces tolerogenic programs and contributes to tumor cell evasion of immune responses. Here, we investigated the relevance of Gal-1 in CRC and explored its modulatory activity within the CD8⁺ regulatory T cell (Treg) compartment. Mice lacking Gal-1 (Lgals1 -/- ) developed a lower number of tumors and showed a decreased frequency of a particular population of CD8⁺CD122⁺PD-1⁺ Tregs in the azoxymethane-dextran sodium sulfate model of colitis-associated CRC. Moreover, silencing of tumor-derived Gal-1 in the syngeneic CT26 CRC model resulted in reduced number and attenuated immunosuppressive capacity of CD8⁺CD122⁺PD-1⁺ Tregs, leading to slower tumor growth. Moreover, stromal Gal-1 also influenced the fitness of CD8⁺ Tregs, highlighting the contribution of both tumor and stromal-derived Gal-1 to this immunoregulatory effect. Finally, bioinformatic analysis of a colorectal adenocarcinoma from The Cancer Genome Atlas dataset revealed a particular signature characterized by high CD8⁺ Treg score and elevated Gal-1 expression, which delineates poor prognosis in human CRC. Our findings identify CD8⁺CD122⁺PD-1⁺ Tregs as a target of the immunoregulatory activity of Gal-1, suggesting a potential immunotherapeutic strategy for the treatment of CRC.Centro de Investigaciones Inmunológicas Básicas y Aplicada

    The non-steroidal anti-inflammatory agent indomethacin differentially controls the regulatory activity of myeloid-derived supressor cells in normal and tumor-associated microenvironments

    No full text
    Las células mieloides supresoras (CMS) constituyen una población de células con actividad regulatoria que controla las respuestas inflamatorias crónicas y promueve el escape tumoral. En este proyecto de tesis mostramos que el efecto del antiinflamatorio no esteroide indometacina (Indo) sobre las CMS es dependiente del contexto en el que se diferencie esta población celular. Ratones BALB/c inoculados con el adenocarcinoma de pulmón singeneico LP07 (3x105 células) se trataron in vivo con una dosis de 10 μM de Indo en el agua de beber. El tratamiento inhibió la acumulación de CMS en el tumor y en órganos linfáticos secundarios producida durante la progresión tumoral, inhibió la actividad supresora de las CMS esplénicas sobre la respuesta linfocitaria a un aloantígeno e inhibió el crecimiento tumoral en un ensayo de transferencia adoptiva, p<0.001. En línea con estos resultados, observamos la misma actividad en las CMS cuando se trataron in vitro con Indo (10 μM) en presencia de medio condicionado de células LP07, simulando el microambiente tumoral. Sin embargo, en ausencia del microambiente tumoral, la administración de Indo generó un aumento en la capacidad supresora (MLR; p< 0.05) y pro-tumoral (transferencia adoptiva, p<0.01) de las CMS. Además, las CMS de ratones normales tratadas con Indo en ausencia del microambiente tumoral suprimieron el desarrollo de la encefalomielitis aguda experimental (EAE), un modelo de esclerosis múltiple en ratones C57BL/6, mientras que las CMS tumorales tratadas con Indo aumentaron la severidad de los síntomas e impidieron su resolución (p<0.05). Analizando los mecanismos de inmunosupresión, Indo redujo la actividad arginasa (4,794 vs 2,077 μg urea/ mg total proteína, p<0.001) y la producción de óxido nítrico (9.2 vs 4.4 μM; P<0,05) y de especies reactivas de oxígeno (124 vs 87 MFI P<0.05) en CMS tumorales pero no en CMS normales. Este estudio revela el rol dual de un agente antiinflamatorio en el control de una población celular regulatoria con implicancias críticas en cáncer y en desórdenes autoinmunes.Myeloid-derived suppressor cells (MDSCs) have emerged as a regulatory cell population that controls chronic inflammatory responses and promotes tumor-immune escape. To date no specific immunomodulatory drug has proven efficacy in targeting the expansion and/or function of these cells. Here we show a context-dependent effect of the non-steroidal antiinflammatory drug indomethacin (Indo) on MDSCs. BALB/c mice were injected with 3x105 LP07 lung adenocarcinoma cells and treated with a non-cytotoxic dose of Indo (10 μM; IC50 LP07: 60 μM). Indo treatment inhibited the suppressive activity of splenic MDSCs in mixed lymphocyte reactions (MLR) (p<0.05), and restrained tumor growth in adoptive transfer experiments (p<0.001). Accordingly, the same effect was observed when MDSCs were treated with Indo in vitro (10 μM; IC50 MDSCs: 125 μM) in the presence of conditioned media from LP07 cells. However, in the absence of a tumoral context, Indo administration increased the suppressive activity of these cells (MLR; p< 0.05) and their pro-tumoral effect (p<0.01). In keeping with these observations, Indo-treated normal MDSCs suppressed the development of Experimental Autoimmune Encephalomyelitis (EAE), a model of multiple sclerosis in C57BL/6 mice, while Indo-treated tumoral MDSCs enhanced the severity of EAE and delayed its resolution (p<0.05). Mechanistically, Indo reduced arginase activity (4,794 vs 2,077 μg urea/ mg total protein,p<0.001) and production of nitric oxide (9.2 vs 4.4 μM; p<0,05) and reactive oxygen species (ROS) (124 vs 87 MFI P<0.05) in tumoral MDSCs, but not in normal MDSCs. Our study unveils the dual role of a widely used anti-inflammatory agent in the control of regulatory cell compartments with critical therapeutic implications in cancer and autoimmune disorders.Fil:Blidner, Ada G.. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales; Argentina

    Cancer immunotherapy–related adverse events: causes and challenges

    No full text
    Despite the success and ongoing promise of monoclonal antibody–targeted immune checkpoint inhibitor immunotherapy of advanced malignancies, in particular, antibodies directed against CTLA-4 and PD-1/PD-L1, the development of immune-related adverse events (irAEs) remains a constraint of this type of therapy. Although rarely fatal, the occurrence of irAEs may necessitate discontinuation of immunotherapy, as well as administration of corticosteroids or other immunosuppressive therapies that may not only compromise efficacy but also predispose for development of opportunistic infection. Clearly, retention of efficacy of immune checkpoint–targeted therapies with concurrent attenuation of immune-mediated toxicity represents a formidable challenge. In this context, the current brief review examines mechanistic relationships between these events, as well as recent insights into immunopathogenesis, and strategies which may contribute to resolving this issue. These sections are preceded by brief overviews of the discovery and functions of CTLA-4 and PD-1, as well as the chronology of the development of immunotherapeutic monoclonal antibodies which target these immune checkpoint inhibitors.Fil: Blidner, Ada Gabriela. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; ArgentinaFil: Choi, Jennifer Ailen. Northwestern University; Estados UnidosFil: Cooksley, Tim. University of Manchester; Reino UnidoFil: Dougan, Michael. Harvard Medical School; Estados UnidosFil: Glezerman, Ilya. Memorial Sloan-kettering Cancer Center.; Estados UnidosFil: Ginex, Pamela. Oncology Nursing Society; Estados UnidosFil: Girotra, Monica. Weill Cornell Medicine; Estados Unidos. Memorial Sloan-kettering Cancer Center.; Estados UnidosFil: Gupta, Dipti. Memorial Sloan-kettering Cancer Center.; Estados UnidosFil: Johnson, Douglas. Vanderbilt University; Estados UnidosFil: Shannon, Vickie R.. University of Texas; Estados UnidosFil: Suarez Almazor, Maria. University of Texas; Estados UnidosFil: Rapoport, Bernardo L.. University of Pretoria; Sudáfrica. Medical Oncology Centre of Rosebank; SudáfricaFil: Anderson, Ronald. University of Pretoria; Sudáfric

    Re-wiring regulatory cell networks in immunity by galectin-glycan interactions

    Get PDF
    Programs that control immune cell homeostasis are orchestrated through the coordinated action of a number of regulatory cell populations, including regulatory T cells, regulatory B cells, myeloid-derived suppressor cells, alternatively-activated macrophages and tolerogenic dendritic cells. These regulatory cell populations can prevent harmful inflammation following completion of protective responses and thwart the development of autoimmune pathology. However, they also have a detrimental role in cancer by favoring escape from immune surveillance. One of the hallmarks of regulatory cells is their remarkable plasticity as they can be positively or negatively modulated by a plethora of cytokines, growth factors and co-stimulatory signals that tailor their differentiation, stability and survival. Here we focus on the emerging roles of galectins, a family of highly conserved glycan-binding proteins in regulating the fate and function of regulatory immune cell populations, both of lymphoid and myeloid origins. Given the broad distribution of circulating and tissue-specific galectins, understanding the relevance of lectin-glycan interactions in shaping regulatory cell compartments will contribute to the design of novel therapeutic strategies aimed at modulating their function in a broad range of immunological disorders.Fil: Blidner, Ada Gabriela. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental (i); ArgentinaFil: Mendez Huergo, Santiago Patricio. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental (i); ArgentinaFil: Cagnoni, Alejandro. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental (i); ArgentinaFil: Rabinovich, Gabriel Adrian. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental (i); Argentina. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales; Argentin
    corecore